101
|
Amand M, Erpicum C, Bajou K, Cerignoli F, Blacher S, Martin M, Dequiedt F, Drion P, Singh P, Zurashvili T, Vandereyken M, Musumeci L, Mustelin T, Moutschen M, Gilles C, Noel A, Rahmouni S. DUSP3/VHR is a pro-angiogenic atypical dual-specificity phosphatase. Mol Cancer 2014; 13:108. [PMID: 24886454 PMCID: PMC4038117 DOI: 10.1186/1476-4598-13-108] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 04/30/2014] [Indexed: 12/20/2022] Open
Abstract
Background DUSP3 phosphatase, also known as Vaccinia-H1 Related (VHR) phosphatase, encoded by DUSP3/Dusp3 gene, is a relatively small member of the dual-specificity protein phosphatases. In vitro studies showed that DUSP3 is a negative regulator of ERK and JNK pathways in several cell lines. On the other hand, DUSP3 is implicated in human cancer. It has been alternatively described as having tumor suppressive and oncogenic properties. Thus, the available data suggest that DUSP3 plays complex and contradictory roles in tumorigenesis that could be cell type-dependent. Since most of these studies were performed using recombinant proteins or in cell-transfection based assays, the physiological function of DUSP3 has remained elusive. Results Using immunohistochemistry on human cervical sections, we observed a strong expression of DUSP3 in endothelial cells (EC) suggesting a contribution for this phosphatase to EC functions. DUSP3 downregulation, using RNA interference, in human EC reduced significantly in vitro tube formation on Matrigel and spheroid angiogenic sprouting. However, this defect was not associated with an altered phosphorylation of the documented in vitro DUSP3 substrates, ERK1/2, JNK1/2 and EGFR but was associated with an increased PKC phosphorylation. To investigate the physiological function of DUSP3, we generated Dusp3-deficient mice by homologous recombination. The obtained DUSP3−/− mice were healthy, fertile, with no spontaneous phenotype and no vascular defect. However, DUSP3 deficiency prevented neo-vascularization of transplanted b-FGF containing Matrigel and LLC xenograft tumors as evidenced by hemoglobin (Hb) and FITC-dextran quantifications. Furthermore, we found that DUSP3 is required for b-FGF-induced microvessel outgrowth in the aortic ring assay. Conclusions All together, our data identify DUSP3 as a new important player in angiogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Souad Rahmouni
- Immunology and Infectious Diseases, GIGA-Signal Transduction, University of Liège, Liège 4000, Belgium.
| |
Collapse
|
102
|
Wong L, Gipp J, Carr J, Loftus C, Benck M, Lee S, Mehta V, Vezina C, Bushman W. Prostate angiogenesis in development and inflammation. Prostate 2014; 74:346-58. [PMID: 24293357 PMCID: PMC3901368 DOI: 10.1002/pros.22751] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/22/2013] [Indexed: 01/07/2023]
Abstract
BACKGROUND Prostatic inflammation is an important factor in development and progression of BPH/LUTS. This study was performed to characterize the normal development and vascular anatomy of the mouse prostate and then examine, for the first time, the effects of prostatic inflammation on the prostate vasculature. METHODS Adult mice were perfused with India ink to visualize the prostatic vascular anatomy. Immunostaining was performed on the E16.5 UGS and the P5, P20, and adult prostate to characterize vascular development. Uropathogenic E. coli 1677 was instilled transurethrally into adult male mice to induce prostate inflammation. RT-PCR and BrdU labeling was performed to assay anigogenic factor expression and endothelial proliferation, respectively. RESULTS An artery on the ventral surface of the bladder trifurcates near the bladder neck to supply the prostate lobes and seminal vesicle. Development of the prostatic vascular system is associated with endothelial proliferation and robust expression of pro-angiogenic factors Pecam1, Tie1, Tek, Angpt1, Angpt2, Fgf2, Vegfa, Vegfc, and Figf. Bacterial-induced prostatic inflammation induced endothelial cell proliferation and increased vascular density but surprisingly decreased pro-angiogenic factor expression. CONCLUSIONS The striking decrease in pro-angiogenic factor mRNA expression associated with endothelial proliferation and increased vascular density during inflammation suggests that endothelial response to injury is not a recapitulation of normal development and may be initiated and regulated by different regulatory mechanisms.
Collapse
Affiliation(s)
- Letitia Wong
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Molecular and Environmental Toxicology Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jerry Gipp
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jason Carr
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Christopher Loftus
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Molly Benck
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Sanghee Lee
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Vatsal Mehta
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Chad Vezina
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Wade Bushman
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
- Correspondence to: Dr. Wade Bushman, MD, PhD, Department of Urology, University of Wisconsin School of Medicine and Public Health, K6/562 Clinical Sciences Center, 600 Highland Avenue, Madison, WI 53792.
| |
Collapse
|
103
|
Voronov E, Carmi Y, Apte RN. The role IL-1 in tumor-mediated angiogenesis. Front Physiol 2014; 5:114. [PMID: 24734023 PMCID: PMC3975103 DOI: 10.3389/fphys.2014.00114] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 03/07/2014] [Indexed: 12/12/2022] Open
Abstract
Tumor angiogenesis is one of the hallmarks of tumor progression and is essential for invasiveness and metastasis. Myeloid inflammatory cells, such as immature myeloid precursor cells, also termed myeloid-derived suppressor cells (MDSCs), neutrophils, and monocytes/macrophages, are recruited to the tumor microenvironment by factors released by the malignant cells that are subsequently “educated” in situ to acquire a pro-invasive, pro-angiogenic, and immunosuppressive phenotype. The proximity of myeloid cells to endothelial cells (ECs) lining blood vessels suggests that they play an important role in the angiogenic response, possibly by secreting a network of cytokines/chemokines and inflammatory mediators, as well as via activation of ECs for proliferation and secretion of pro-angiogenic factors. Interleukin-1 (IL-1) is an “alarm,” upstream, pro-inflammatory cytokine that is generated primarily by myeloid cells. IL-1 initiates and propagates inflammation, mainly by inducing a local cytokine network and enhancing inflammatory cell infiltration to affected sites and by augmenting adhesion molecule expression on ECs and leukocytes. Pro-inflammatory mediators were recently shown to play an important role in tumor-mediated angiogenesis and blocking their function may suppress tumor progression. In this review, we summarize the interactions between IL-1 and other pro-angiogenic factors during normal and pathological conditions. In addition, the feasibility of IL-1 neutralization approaches for anti-cancer therapy is discussed.
Collapse
Affiliation(s)
- Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | - Yaron Carmi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | - Ron N Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev Beer-Sheva, Israel
| |
Collapse
|
104
|
Dutcher JP, Neuberg D, Atkins MB, Tester WJ, Wadler S, Stewart JA, Chachoua A, Schuchter LM. Report of a phase I evaluation of dose and schedule of interleukin-1 alpha and cyclophosphamide in patients with advanced tumors: An Eastern Cooperative Oncology Group study (PX990) and review of IL-1-based studies of hematopoietic reconstitution. J Interferon Cytokine Res 2014; 34:376-84. [PMID: 24433038 DOI: 10.1089/jir.2013.0010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Interleukin-1 (IL-1) is a cytokine critical to inflammation, immunological activation, response to infection, and bone marrow hematopoiesis. Cyclophosphamide downmodulates immune suppressor cells and is cytotoxic to a variety of tumors. A phase I trial of IL-1 and cyclophosphamide was conducted by the Eastern Cooperative Oncology Group. This study evaluated 3 dose levels and 3 schedules in patients with solid tumors. The goal was to evaluate the hematopoietic supportive care effect and possible antitumor effect. Toxicity was fever, chills, hypotension, nausea/emesis, hepatic, and neutropenia. Toxicity increased with dose increases of interleukin-1. Treatment at all dose levels resulted in significant increases in total white blood cell (WBC) counts above baseline. Nadir WBC and nadir absolute neutrophil counts were not significantly different by dose level of IL-1 or schedule of IL-1. Toxicity due to IL-1 at higher doses prohibited further evaluation of this agent for hematopoietic support, particularly in view of the activity and tolerability of more lineage-specific hematopoietic cytokines. Therapeutic interventions in the role of IL-1 in inflammatory conditions and cancer may be further informed by our definition of its clinical and biological effects in this evaluation of dose and schedule.
Collapse
Affiliation(s)
- Janice P Dutcher
- 1 Department of Oncology, Montefiore Medical Center , Bronx, New York
| | | | | | | | | | | | | | | |
Collapse
|
105
|
Pereira MSS, Rossi MA, Cardoso CR, da Silva JS, Bezerra da Silva LA, Kuga MC, Faria G. Cellular and molecular tissue response to triple antibiotic intracanal dressing. J Endod 2014; 40:499-504. [PMID: 24666899 DOI: 10.1016/j.joen.2013.10.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 09/26/2013] [Accepted: 10/18/2013] [Indexed: 02/09/2023]
Abstract
INTRODUCTION The aim of this study was to characterize the response of mouse subcutaneous tissue to triple antibiotic paste (TAP) using conventional light microscopy and real-time PCR (qRT-PCR). METHODS Polyethylene tubes containing TAP or calcium hydroxide (CH) (ie, the control group) were implanted in mouse subcutaneous tissue. Animals that received empty tubes or no tubes were used as additional controls. After periods of 7, 21, and 63 days postimplantation, the specimens were removed and subjected to histologic processing. The number of inflammatory cells and vessels, vessel areas, vascular density, and relative percentage of collagen were evaluated. Gene expression of proinflammatory (interleukin-1 beta, tumor necrosis factor alpha, and interleukin 17) and anti-inflammatory (transforming growth factor beta) cytokines and angiogenic factors (vascular endothelial growth factor and hypoxia-inducible factor-1 alpha) was quantified by 7 and 21 days postimplantation. Results were analyzed using the Student t test, analysis of variance, and the Tukey test (α = 0.05). RESULTS TAP induced an exuberant inflammatory and angiogenic response, with higher numbers of inflammatory cells, higher vascular area and density, and lower relative percentage of collagen compared with CH. In general, the expression of genes involved in inflammation and angiogenesis was higher in the TAP group compared with animals that received CH or empty tubes. CONCLUSIONS The response of mouse subcutaneous tissue to TAP was characterized by exuberant and persistent inflammatory and angiogenic responses with no repair and high gene expression of biomarkers associated with inflammation and angiogenesis.
Collapse
Affiliation(s)
- Maristela Soares Swerts Pereira
- Department of Paediatric Dentistry, Preventive and Community Dentistry, Ribeirão Preto Dentistry School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Paediatric Dentistry, Alfenas Dentistry School, University José Rosário Vellano, Unifenas, Alfenas, Minas Gerais, Brazil
| | | | - Cristina Ribeiro Cardoso
- Department of Clinical Analyzes, Toxixology and Food Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - João Santana da Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Léa Assed Bezerra da Silva
- Department of Paediatric Dentistry, Preventive and Community Dentistry, Ribeirão Preto Dentistry School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Milton Carlos Kuga
- Department of Restorative Dentistry, Araraquara Dental School, UNESP - Univ Estadual Paulista, Araraquara, São Paulo, Brazil
| | - Gisele Faria
- Department of Restorative Dentistry, Araraquara Dental School, UNESP - Univ Estadual Paulista, Araraquara, São Paulo, Brazil.
| |
Collapse
|
106
|
Arango Duque G, Descoteaux A. Macrophage cytokines: involvement in immunity and infectious diseases. Front Immunol 2014; 5:491. [PMID: 25339958 PMCID: PMC4188125 DOI: 10.3389/fimmu.2014.00491] [Citation(s) in RCA: 1411] [Impact Index Per Article: 141.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 09/22/2014] [Indexed: 12/21/2022] Open
Abstract
The evolution of macrophages has made them primordial for both development and immunity. Their functions range from the shaping of body plans to the ingestion and elimination of apoptotic cells and pathogens. Cytokines are small soluble proteins that confer instructions and mediate communication among immune and non-immune cells. A portfolio of cytokines is central to the role of macrophages as sentries of the innate immune system that mediate the transition from innate to adaptive immunity. In concert with other mediators, cytokines bias the fate of macrophages into a spectrum of inflammation-promoting "classically activated," to anti-inflammatory or "alternatively activated" macrophages. Deregulated cytokine secretion is implicated in several disease states ranging from chronic inflammation to allergy. Macrophages release cytokines via a series of beautifully orchestrated pathways that are spatiotemporally regulated. At the molecular level, these exocytic cytokine secretion pathways are coordinated by multi-protein complexes that guide cytokines from their point of synthesis to their ports of exit into the extracellular milieu. These trafficking proteins, many of which were discovered in yeast and commemorated in the 2013 Nobel Prize in Physiology or Medicine, coordinate the organelle fusion steps that are responsible for cytokine release. This review discusses the functions of cytokines secreted by macrophages, and summarizes what is known about their release mechanisms. This information will be used to delve into how selected pathogens subvert cytokine release for their own survival.
Collapse
Affiliation(s)
- Guillermo Arango Duque
- INRS-Institut Armand-Frappier, Laval, QC, Canada
- Centre for Host-Parasite Interactions, Laval, QC, Canada
- *Correspondence: Guillermo Arango Duque and Albert Descoteaux, Institut National de la Recherche Scientifique–Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada e-mail: , ;
| | - Albert Descoteaux
- INRS-Institut Armand-Frappier, Laval, QC, Canada
- Centre for Host-Parasite Interactions, Laval, QC, Canada
- *Correspondence: Guillermo Arango Duque and Albert Descoteaux, Institut National de la Recherche Scientifique–Institut Armand-Frappier, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada e-mail: , ;
| |
Collapse
|
107
|
Silvestre JS, Smadja DM, Lévy BI. Postischemic revascularization: from cellular and molecular mechanisms to clinical applications. Physiol Rev 2013; 93:1743-802. [PMID: 24137021 DOI: 10.1152/physrev.00006.2013] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
After the onset of ischemia, cardiac or skeletal muscle undergoes a continuum of molecular, cellular, and extracellular responses that determine the function and the remodeling of the ischemic tissue. Hypoxia-related pathways, immunoinflammatory balance, circulating or local vascular progenitor cells, as well as changes in hemodynamical forces within vascular wall trigger all the processes regulating vascular homeostasis, including vasculogenesis, angiogenesis, arteriogenesis, and collateral growth, which act in concert to establish a functional vascular network in ischemic zones. In patients with ischemic diseases, most of the cellular (mainly those involving bone marrow-derived cells and local stem/progenitor cells) and molecular mechanisms involved in the activation of vessel growth and vascular remodeling are markedly impaired by the deleterious microenvironment characterized by fibrosis, inflammation, hypoperfusion, and inhibition of endogenous angiogenic and regenerative programs. Furthermore, cardiovascular risk factors, including diabetes, hypercholesterolemia, hypertension, diabetes, and aging, constitute a deleterious macroenvironment that participates to the abrogation of postischemic revascularization and tissue regeneration observed in these patient populations. Thus stimulation of vessel growth and/or remodeling has emerged as a new therapeutic option in patients with ischemic diseases. Many strategies of therapeutic revascularization, based on the administration of growth factors or stem/progenitor cells from diverse sources, have been proposed and are currently tested in patients with peripheral arterial disease or cardiac diseases. This review provides an overview from our current knowledge regarding molecular and cellular mechanisms involved in postischemic revascularization, as well as advances in the clinical application of such strategies of therapeutic revascularization.
Collapse
|
108
|
Nold-Petry CA, Rudloff I, Baumer Y, Ruvo M, Marasco D, Botti P, Farkas L, Cho SX, Zepp JA, Azam T, Dinkel H, Palmer BE, Boisvert WA, Cool CD, Taraseviciene-Stewart L, Heinhuis B, Joosten LAB, Dinarello CA, Voelkel NF, Nold MF. IL-32 promotes angiogenesis. THE JOURNAL OF IMMUNOLOGY 2013; 192:589-602. [PMID: 24337385 DOI: 10.4049/jimmunol.1202802] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-32 is a multifaceted cytokine with a role in infections, autoimmune diseases, and cancer, and it exerts diverse functions, including aggravation of inflammation and inhibition of virus propagation. We previously identified IL-32 as a critical regulator of endothelial cell (EC) functions, and we now reveal that IL-32 also possesses angiogenic properties. The hyperproliferative ECs of human pulmonary arterial hypertension and glioblastoma multiforme exhibited a markedly increased abundance of IL-32, and, significantly, the cytokine colocalized with integrin αVβ3. Vascular endothelial growth factor (VEGF) receptor blockade, which resulted in EC hyperproliferation, increased IL-32 three-fold. Small interfering RNA-mediated silencing of IL-32 negated the 58% proliferation of ECs that occurred within 24 h in scrambled-transfected controls. Reduction of IL-32 neither affected apoptosis (insignificant changes in Bak-1, Bcl-2, Bcl-xL, lactate dehydrogenase, annexin V, and propidium iodide) nor VEGF or TGF-β levels, but siIL-32-transfected adult and neonatal ECs produced up to 61% less NO, IL-8, and matrix metalloproteinase-9, and up to 3-fold more activin A and endostatin. In coculture-based angiogenesis assays, IL-32γ dose-dependently increased tube formation up to 3-fold; an αVβ3 inhibitor prevented this activity and reduced IL-32γ-induced IL-8 by 85%. In matrigel plugs loaded with IL-32γ, VEGF, or vehicle and injected into live mice, we observed the anticipated VEGF-induced increase in neocapillarization (8-fold versus vehicle), but unexpectedly, IL-32γ was equally angiogenic. A second signal such as IFN-γ was required to render cells responsive to exogenous IL-32γ; importantly, this was confirmed using a completely synthetic preparation of IL-32γ. In summary, we add angiogenic properties that are mediated by integrin αVβ3 but VEGF-independent to the portfolio of IL-32, implicating a role for this versatile cytokine in pulmonary arterial hypertension and neoplastic diseases.
Collapse
Affiliation(s)
- Claudia A Nold-Petry
- Ritchie Centre, Monash Institute of Medical Research, Monash University, Melbourne, Victoria 3168, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Long KB, Beatty GL. Harnessing the antitumor potential of macrophages for cancer immunotherapy. Oncoimmunology 2013; 2:e26860. [PMID: 24498559 PMCID: PMC3902119 DOI: 10.4161/onci.26860] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/15/2013] [Accepted: 10/17/2013] [Indexed: 01/04/2023] Open
Abstract
Macrophages constitute a dominant fraction of the population of immune cells that infiltrate developing tumors. Recruited by tumor-derived signals, tumor-infiltrating macrophages are key orchestrators of a microenvironment that supports tumor progression. However, the phenotype of macrophages is pliable and, if instructed properly, macrophages can mediate robust antitumor functions through their ability to eliminate malignant cells, inhibit angiogenesis, and deplete fibrosis. While much effort has focused on strategies to block the tumor-supporting activity of macrophages, emerging approaches designed to instruct macrophages with antitumor properties are demonstrating promise and may offer a novel strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Kristen B Long
- Abramson Cancer Center; Department of Medicine; Division of Hematology-Oncology; University of Pennsylvania Perelman School of Medicine; Philadelphia, PA USA
| | - Gregory L Beatty
- Abramson Cancer Center; Department of Medicine; Division of Hematology-Oncology; University of Pennsylvania Perelman School of Medicine; Philadelphia, PA USA
| |
Collapse
|
110
|
Abstract
Although the IL-1α molecule has long been recognized, information about its distinct role in various diseases is limited, since most clinical studies have focused on the role of IL-1β. Despite triggering the same IL-1 receptor as does IL-1β, there is, however, a distinct role for IL-1α in some inflammatory diseases. IL-1α is a unique cytokine since it is constitutively present intracellularly in nearly all resting non-hematopoietic cells in health as well as being up-regulated during hypoxia. During cell necrosis, IL-1α functions as an alarm molecule and thus plays a critical role early in inflammation. Following its release from damage tissue cells, IL-1α mediates neutrophil recruitment to the site of injury, inducing IL-1β, other cytokines and chemokines from surrounding resident cells. Another unique attribute of IL-1α is its nuclear localization sequence present in the N-terminal half of the precursor termed the propiece. The IL-1α propiece translocates into the nucleus and participates in the regulation of transcription. Therefore, IL-1α, like IL-1 family members IL-33 and IL-37, is a 'dual-function' cytokine binding to chromatin as well as to its cell surface receptor. Some cancer cells can express membrane IL-1α, which can increase immunogenicity of tumor cells and serve in anti-tumor immune surveillance and tumor regression. However, in the tumor microenvironment, precursor IL-1α released from dying tumor cells is inflammatory and, similar to IL-1β, increases tumor invasiveness and angiogenesis.
Collapse
Affiliation(s)
- Peleg Rider
- Faculty of Health Sciences, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Yaron Carmi
- School of Medicine, Department of Pathology, Stanford University, Palo Alto, CA, USA
| | - Elena Voronov
- The Shraga Segal Department of Microbiology, Immunology and Genetics and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ron N Apte
- The Shraga Segal Department of Microbiology, Immunology and Genetics and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
111
|
Murakami Y, Watari K, Shibata T, Uba M, Ureshino H, Kawahara A, Abe H, Izumi H, Mukaida N, Kuwano M, Ono M. N-myc downstream-regulated gene 1 promotes tumor inflammatory angiogenesis through JNK activation and autocrine loop of interleukin-1α by human gastric cancer cells. J Biol Chem 2013; 288:25025-25037. [PMID: 23846687 DOI: 10.1074/jbc.m113.472068] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The expression of N-myc downstream-regulated gene 1 (NDRG1) was significantly correlated with tumor angiogenesis and malignant progression together with poor prognosis in gastric cancer. However, the underlying mechanism for the role of NDRG1 in the malignant progression of gastric cancer remains unknown. Here we examined whether and how NDRG1 could modulate tumor angiogenesis by human gastric cancer cells. We established NU/Cap12 and NU/Cap32 cells overexpressing NDRG1 in NUGC-3 cells, which show lower tumor angiogenesis in vivo. Compared with parental NU/Mock3, NU/Cap12, and NU/Cap32 cells: 1) induced higher tumor angiogenesis than NU/Mock3 cells accompanied by infiltration of tumor-associated macrophages in mouse dorsal air sac assay and Matrigel plug assay; 2) showed much higher expression of CXC chemokines, MMP-1, and the potent angiogenic factor VEGF-A; 3) increased the expression of the representative inflammatory cytokine, IL-1α; 4) augmented JNK phosphorylation and nuclear expression of activator protein 1 (AP-1). Further analysis demonstrated that knockdown of AP-1 (Jun and/or Fos) resulted in down-regulation of the expression of VEGF-A, CXC chemokines, and MMP-1, and also suppressed expression of IL-1α in NDRG1-overexpressing cell lines. Treatment with IL-1 receptor antagonist (IL-1ra) resulted in down-regulation of JNK and c-Jun phosphorylation, and the expression of VEGF-A, CXC chemokines, and MMP-1 in NU/Cap12 and NU/Cap32 cells. Finally, administration of IL-1ra suppressed both tumor angiogenesis and infiltration of macrophages by NU/Cap12 in vivo. Together, activation of JNK/AP-1 thus seems to promote tumor angiogenesis in relationship to NDRG1-induced inflammatory stimuli by gastric cancer cells.
Collapse
Affiliation(s)
- Yuichi Murakami
- From the Department of Pharmaceutical Oncology and; the St. Mary's Hospital, Kurume 830-8543
| | | | | | - Manami Uba
- From the Department of Pharmaceutical Oncology and
| | | | - Akihiko Kawahara
- the Department of Diagnostic Pathology, Kurume University Hospital, Kurume 830-0011
| | - Hideyuki Abe
- the Department of Diagnostic Pathology, Kurume University Hospital, Kurume 830-0011
| | - Hiroto Izumi
- the Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Kitakyushu 807-8555, and
| | - Naofumi Mukaida
- the Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-0934, Japan
| | - Michihiko Kuwano
- Laboratory of Molecular Cancer Biology, Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582
| | - Mayumi Ono
- From the Department of Pharmaceutical Oncology and.
| |
Collapse
|
112
|
van de Veerdonk FL, Netea MG. New Insights in the Immunobiology of IL-1 Family Members. Front Immunol 2013; 4:167. [PMID: 23847614 PMCID: PMC3703542 DOI: 10.3389/fimmu.2013.00167] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 06/12/2013] [Indexed: 12/28/2022] Open
Abstract
The interleukin-1 (IL 1) family of ligands is associated with acute and chronic inflammation, and plays an essential role in the non-specific innate response to infection. The biological properties of IL 1 family ligands are typically pro-inflammatory. The IL 1 family has 11 family members and can be categorized into subfamilies according to the length of their precursor and the length of the propiece for each precursor (Figure 1). The IL 1 subfamily consists of IL 1α, IL 1β, and IL 33, with the longest propieces of the IL 1 family. IL 18 and IL 37 belong to the IL 18 subfamily and contain smaller propieces than IL 1 and IL-33. Since IL 37 binds to the IL 18Rα chain it is part of the IL 18 subfamily, however it remains to be elucidated how the propiece of IL 37 is removed. IL 36α, β, and γ as well as IL 36 Ra belong to the IL 36 subfamily. In addition, IL 38 likely belongs to this family since it has the ability to bind to the IL 36R. The IL 36 subfamily has the shortest propiece. The one member of the IL 1 family that cannot be categorized in these subfamilies is IL 1 receptor antagonist (IL 1Ra), which has a signal peptide and is readily secreted. In the present review we will describe the biological functions of the IL-1F members and new insights in their biology.
Collapse
Affiliation(s)
- Frank L van de Veerdonk
- Department of Medicine, Radboud University Nijmegen Medical Center, Nijmegen Institute for Infection, Inflammation and Immunity (N4i) , Nijmegen , Netherlands
| | | |
Collapse
|
113
|
Rivera JC, Sitaras N, Noueihed B, Hamel D, Madaan A, Zhou T, Honoré JC, Quiniou C, Joyal JS, Hardy P, Sennlaub F, Lubell W, Chemtob S. Microglia and interleukin-1β in ischemic retinopathy elicit microvascular degeneration through neuronal semaphorin-3A. Arterioscler Thromb Vasc Biol 2013; 33:1881-91. [PMID: 23766263 DOI: 10.1161/atvbaha.113.301331] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Proinflammatory cytokines contribute to the development of retinal vasculopathies. However, the role of these factors and the mechanisms by which they elicit their effects in retina are not known. We investigated whether activated microglia during early stages of ischemic retinopathy produces excessive interleukin-1β (IL-1β), which elicits retinal microvascular degeneration not directly but rather by triggering the release of the proapoptotic/repulsive factor semaphorin-3A (Sema3A) from neurons. APPROACH AND RESULTS Sprague Dawley rats subjected to retinopathy induced by hyperoxia (80% O2; O2-induced retinopathy) exhibited retinal vaso-obliteration associated with microglial activation, NLRP3 upregulation, and IL-1β and Sema3A release; IL-1β was mostly generated by microglia. Intraperitoneal administration of IL-1 receptor antagonists (Kineret, or rytvela [101.10]) decreased these effects and enhanced retinal revascularization; knockdown of Sema3A resulted in microvessel preservation and, conversely, administration of IL-1β caused vaso-obliteration. In vitro, IL-1β derived from activated primary microglial cells, cultured under hyperoxia, stimulated the release of Sema3A in retinal ganglion cells-5, which in turn induced apoptosis of microvascular endothelium; antagonism of IL-1 receptor decreased microglial activation and on retinal ganglion cells-5 abolished the release of Sema3A inhibiting ensuing endothelial cell apoptosis. IL-1β was not directly cytotoxic to endothelial cells. CONCLUSIONS Our findings suggest that in the early stages of O2-induced retinopathy, retinal microglia are activated to produce IL-1β, which sustains the activation of microglia and induces microvascular injury through the release of Sema3A from adjacent neurons. Interference with IL-1 receptor or Sema3A actions preserves the microvascular bed in ischemic retinopathies and, consequently, decreases ensued pathological preretinal neovascularization.
Collapse
Affiliation(s)
- José Carlos Rivera
- Department of Pediatrics, Ophthalmology, and Pharmacology, CHU Sainte-Justine Research Center, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Maruotti N, Annese T, Cantatore FP, Ribatti D. Macrophages and angiogenesis in rheumatic diseases. Vasc Cell 2013; 5:11. [PMID: 23725043 PMCID: PMC3680215 DOI: 10.1186/2045-824x-5-11] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 04/26/2013] [Indexed: 12/31/2022] Open
Abstract
Angiogenesis plays a key role in several rheumatic diseases, including rheumatoid arthritis, osteoarthritis, ankylosing spondylitis, systemic sclerosis, systemic lupus erythematosus, and vasculitides. An imbalance between angiogenic inducers and inhibitors seems to be a critical factor in pathogenesis of these diseases. Macrophages promote angiogenesis during rheumatoid arthritis. In addition, macrophages can produce a variety of pro-angiogenic factors that have been associated with the angiogenic response occurring during other rheumatic diseases. Lastly, macrophages could be a target in the treatment of rheumatoid arthritis and other rheumatic diseases. Nevertheless, further studies are needed to better elucidate the exact role of macrophage in angiogenesis in these diseases.
Collapse
Affiliation(s)
- Nicola Maruotti
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia Medical School- Ospedale “ D’Avanzo”, Foggia, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Piazza Giulio Cesare, 11, Policlinico, 70124, Bari, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia Medical School- Ospedale “ D’Avanzo”, Foggia, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Piazza Giulio Cesare, 11, Policlinico, 70124, Bari, Italy
| |
Collapse
|
115
|
Carmi Y, Dotan S, Rider P, Kaplanov I, White MR, Baron R, Abutbul S, Huszar M, Dinarello CA, Apte RN, Voronov E. The role of IL-1β in the early tumor cell-induced angiogenic response. THE JOURNAL OF IMMUNOLOGY 2013; 190:3500-9. [PMID: 23475218 DOI: 10.4049/jimmunol.1202769] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this study, we assessed the involvement of IL-1β in early angiogenic responses induced by malignant cells using Matrigel plugs supplemented with B16 melanoma cells. We found that during the angiogenic response, IL-1β and vascular endothelial growth factor (VEGF) interact in a newly described autoinduction circuit, in which each of these cytokines induces the other. The IL-1β and VEGF circuit acts through interactions between bone marrow-derived VEGF receptor 1(+)/IL-1R1(+) immature myeloid cells and tissue endothelial cells. Myeloid cells produce IL-1β and additional proinflammatory cytokines, which subsequently activate endothelial cells to produce VEGF and other proangiogenic factors and provide the inflammatory microenvironment for angiogenesis and tumor progression. These mechanisms were also observed in a nontumor early angiogenic response elicited in Matrigel plugs by either rIL-1β or recombinant VEGF. We have shown that IL-1β inhibition stably reduces tumor growth by limiting inflammation and inducing the maturation of immature myeloid cells into M1 macrophages. In sharp contrast, only transient inhibition of tumor growth was observed after VEGF neutralization, followed by tumor recurrence mediated by rebound angiogenesis. This occurs via the reprogramming of VEGF receptor 1(+)/IL-1R1(+) cells to express hypoxia inducible factor-1α, VEGF, and other angiogenic factors, thereby directly supporting proliferation of endothelial cells and blood vessel formation in a paracrine manner. We suggest using IL-1β inhibition as an effective antitumor therapy and are currently optimizing the conditions for its application in the clinic.
Collapse
Affiliation(s)
- Yaron Carmi
- The Shraga Segal Department of Microbiology, Immunology and Genetics and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Baluk P, Hogmalm A, Bry M, Alitalo K, Bry K, McDonald DM. Transgenic overexpression of interleukin-1β induces persistent lymphangiogenesis but not angiogenesis in mouse airways. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1434-47. [PMID: 23391392 DOI: 10.1016/j.ajpath.2012.12.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 12/21/2012] [Accepted: 12/31/2012] [Indexed: 01/03/2023]
Abstract
These studies used bi-transgenic Clara cell secretory protein (CCSP)/IL-1β mice that conditionally overexpress IL-1β in Clara cells to determine whether IL-1β can promote angiogenesis and lymphangiogenesis in airways. Doxycycline treatment induced rapid, abundant, and reversible IL-1β production, influx of neutrophils and macrophages, and conspicuous and persistent lymphangiogenesis, but surprisingly no angiogenesis. Gene profiling showed many up-regulated genes, including chemokines (Cxcl1, Ccl7), cytokines (tumor necrosis factor α, IL-1β, and lymphotoxin-β), and leukocyte genes (S100A9, Aif1/Iba1). Newly formed lymphatics persisted after IL-1β overexpression was stopped. Further studies examined how IL1R1 receptor activation by IL-1β induced lymphangiogenesis. Inactivation of vascular endothelial growth factor (VEGF)-C and VEGF-D by adeno-associated viral vector-mediated soluble VEGFR-3 (VEGF-C/D Trap) completely blocked lymphangiogenesis, showing its dependence on VEGFR-3 ligands. Consistent with this mechanism, VEGF-C immunoreactivity was present in some Aif1/Iba1-immunoreactive macrophages. Because neutrophils contribute to IL-1β-induced lung remodeling in newborn mice, we examined their potential role in lymphangiogenesis. Triple-transgenic CCSP/IL-1β/CXCR2(-/-) mice had the usual IL-1β-mediated lymphangiogenesis but no neutrophil recruitment, suggesting that neutrophils are not essential. IL1R1 immunoreactivity was found on some epithelial basal cells and neuroendocrine cells, suggesting that these cells are targets of IL-1β, but was not detected on lymphatics, blood vessels, or leukocytes. We conclude that lymphangiogenesis triggered by IL-1β overexpression in mouse airways is driven by VEGF-C/D from macrophages, but not neutrophils, recruited by chemokines from epithelial cells that express IL1R1.
Collapse
Affiliation(s)
- Peter Baluk
- Cardiovascular Research Institute, University of California, San Francisco, California 94143-0130, USA.
| | | | | | | | | | | |
Collapse
|
117
|
Pollheimer J, Bodin J, Sundnes O, Edelmann RJ, Skånland SS, Sponheim J, Brox MJ, Sundlisæter E, Loos T, Vatn M, Kasprzycka M, Wang J, Küchler AM, Taskén K, Haraldsen G, Hol J. Interleukin-33 Drives a Proinflammatory Endothelial Activation That Selectively Targets Nonquiescent Cells. Arterioscler Thromb Vasc Biol 2013; 33:e47-55. [DOI: 10.1161/atvbaha.112.253427] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jürgen Pollheimer
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Johanna Bodin
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Olav Sundnes
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Reidunn J. Edelmann
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Sigrid S. Skånland
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Jon Sponheim
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Mari Johanna Brox
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Eirik Sundlisæter
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Tamara Loos
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Morten Vatn
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Monika Kasprzycka
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Junbai Wang
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Axel M. Küchler
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Kjetil Taskén
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Guttorm Haraldsen
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| | - Johanna Hol
- From the LIIPAT, Institute of Pathology, University of Oslo, Oslo, Norway (J.P., O.S., R.J.E., J.S., M.J.B., E.S., T.L., M.K., A.M.K., G.H., J.H.); Department of Pathology, Oslo University Hospital, Oslo, Norway (J.P., J.B., O.S., R.J.E., E.S., T.L., M.K., J.W., A.M.K., G.H., J.H.); Department of Obstetrics and Fetal-Maternal Medicine, Medical University of Vienna, Austria (J.P.); Division of Environmental Medicine, Norwegian Institute of Public Health, Oslo, Norway (J.B.); Centre for Molecular
| |
Collapse
|
118
|
Duda DG. Molecular Biomarkers of Response to Antiangiogenic Therapy for Cancer. ACTA ACUST UNITED AC 2012; 2012. [PMID: 24340224 DOI: 10.5402/2012/587259] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Antiangiogenic therapy for cancer has gone from an intriguing hypothesis in the 1970s to an accepted treatment approach for many cancer types. It has also become a standard of care for certain eye diseases. Yet, despite the use of molecularly targeted drugs with well defined targets, to date there are no biomarkers to guide the use of antiangiogenic therapy in patients. The mechanisms of action of these drugs are also being debated. This paper discusses some of the emerging biomarker candidates for this type of cancer therapy, which have provided mechanistic insight and might be useful in the future for optimizing cancer treatment.
Collapse
Affiliation(s)
- Dan G Duda
- Steele Laboratory for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
119
|
Umansky V, Sevko A. Tumor microenvironment and myeloid-derived suppressor cells. CANCER MICROENVIRONMENT 2012; 6:169-77. [PMID: 23242672 DOI: 10.1007/s12307-012-0126-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 11/14/2012] [Indexed: 01/04/2023]
Abstract
Tumor progression has been demonstrated to be supported by chronic inflammatory conditions developed in the tumor microenvironment and characterized by the long-term secretion of various inflammatory soluble factors (including cytokines, chemokines, growth factors, reactive oxygen and nitrogen species, prostaglandins etc.) and strong leukocyte infiltration. Among leukocytes infiltrating tumors, myeloid-derived suppressor cells (MDSCs) represent one of the most important players mediating immunosuppression. These cells may not only strongly inhibit an anti-tumor immune reactions mediated by T cells but also directly stimulate tumorigenesis, tumor growth and metastasis by enhancing neoangiogenesis and creating a suitable environment for the metastatic formation. This review provides an overview of interactions between MDSCs and tumor cells leading to MDSC generation, activation and migration to the tumor site, where they can strongly enhance tumor progression. Better understanding of the MDSC-tumor interplay is critical for the development of new strategies of tumor immunotherapy.
Collapse
Affiliation(s)
- Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center, 69120, Heidelberg, Germany,
| | | |
Collapse
|
120
|
Rider P, Kaplanov I, Romzova M, Bernardis L, Braiman A, Voronov E, Apte RN. The transcription of the alarmin cytokine interleukin-1 alpha is controlled by hypoxia inducible factors 1 and 2 alpha in hypoxic cells. Front Immunol 2012; 3:290. [PMID: 23049530 PMCID: PMC3442192 DOI: 10.3389/fimmu.2012.00290] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 08/29/2012] [Indexed: 02/04/2023] Open
Abstract
During hypoxia, cells undergo transcriptional changes to adjust to metabolic stress, to promote cell survival, and to induce pro-angiogenic factors. Hypoxia-induced factors (HIFs) regulate these transcriptional alterations. Failure to restore oxygen levels results in cell death by necrosis. IL-1α is one of the most important mediators of sterile inflammation following hypoxia-mediated necrosis. During hypoxia, IL-1α is up-regulated and released from necrotic cells, promoting the initiation of sterile inflammation. This study examined the role of IL-1α transcription in initiation of hypoxic stress and the correlation between IL-1α transcription and HIFα factors. In an epithelial cell line cultured under hypoxic conditions, IL-1α transcription was up-regulated in a process mediated and promoted by HIFα factors. IL-1α transcription was also up-regulated in hypoxia in a fibroblast cell line, however, in these cells, HIFα factors inhibited the elevation of transcription. These data suggest that HIFα factors play a significant role in initiating sterile inflammation by controlling IL-1α transcription during hypoxia in a differential manner, depending on the cell type.
Collapse
Affiliation(s)
- Peleg Rider
- The Shraga Segal Department of Microbiology and Immunology and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev Beer-Sheva, Israel
| | | | | | | | | | | | | |
Collapse
|
121
|
Delta-like 4 inhibits choroidal neovascularization despite opposing effects on vascular endothelium and macrophages. Angiogenesis 2012; 15:609-22. [PMID: 22869002 PMCID: PMC3496480 DOI: 10.1007/s10456-012-9290-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 07/13/2012] [Indexed: 12/11/2022]
Abstract
Inflammatory neovascularization, such as choroidal neovascularization (CNV), occur in the presence of Notch expressing macrophages. DLL4s anti-angiogenic effect on endothelial cells (EC) has been widely recognized, but its influence on Notch signaling on macrophages and its overall effect in inflammatory neovascularization is not well understood. We identified macrophages and ECs as the main Notch 1 and Notch 4 expressing cells in CNV. A soluble fraction spanning Ser28-Pro525 of the murine extracellular DLL4 domain (sDLL4/28-525) activated the Notch pathway, as it induces Notch target genes in macrophages and ECs and inhibited EC proliferation and vascular sprouting in aortic rings. In contrast, sDLL4/28-525 increased pro-angiogenic VEGF, and IL-1β expression in macrophages responsible for increased vascular sprouting observed in aortic rings incubated in conditioned media from sDLL4/28-525 stimulated macrophages. In vivo, Dll4+/− mice developed significantly more CNV and sDLL4/28-525 injections inhibited CNV in Dll4+/− CD1 mice. Similarly, sDLL4/28-525 inhibited CNV in C57Bl6 and its effect was reversed by a γ-secretase inhibitor that blocks Notch signaling. The inhibition occurred despite increased VEGF, IL-1β expression in infiltrating inflammatory macrophages in sDLL4/28-525 treated mice and might be due to direct inhibition of EC proliferation in laser-induced CNV as demonstrated by EdU labelling in vivo. In conclusion, Notch activation on macrophages and ECs leads to opposing effects in inflammatory neovascularization in situations such as CNV.
Collapse
|
122
|
Dinarello CA, Simon A, van der Meer JWM. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov 2012; 11:633-52. [PMID: 22850787 PMCID: PMC3644509 DOI: 10.1038/nrd3800] [Citation(s) in RCA: 1283] [Impact Index Per Article: 106.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interleukin-1 (IL-1) is a highly active pro-inflammatory cytokine that lowers pain thresholds and damages tissues. Monotherapy blocking IL-1 activity in autoinflammatory syndromes results in a rapid and sustained reduction in disease severity, including reversal of inflammation-mediated loss of sight, hearing and organ function. This approach can therefore be effective in treating common conditions such as post-infarction heart failure, and trials targeting a broad spectrum of new indications are underway. So far, three IL-1-targeted agents have been approved: the IL-1 receptor antagonist anakinra, the soluble decoy receptor rilonacept and the neutralizing monoclonal anti-IL-1β antibody canakinumab. In addition, a monoclonal antibody directed against the IL-1 receptor and a neutralizing anti-IL-1α antibody are in clinical trials.
Collapse
Affiliation(s)
- Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, Colorado 80045, USA.
| | | | | |
Collapse
|
123
|
Shih KS, Wang JH, Wu YW, Teng CM, Chen CC, Yang CR. Aciculatin inhibits granulocyte colony-stimulating factor production by human interleukin 1β-stimulated fibroblast-like synoviocytes. PLoS One 2012; 7:e42389. [PMID: 22860122 PMCID: PMC3409160 DOI: 10.1371/journal.pone.0042389] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 07/04/2012] [Indexed: 01/25/2023] Open
Abstract
The expression of granulocyte colony-stimulating factor (G-CSF), the major regulator of neutrophil maturation, by human fibroblast-like synoviocytes (FLS) can be stimulated by the inflammatory cytokine interleukin-1β (IL-1β). G-CSF is known to contribute to the pathologic processes of destructive arthritis, but the induction mechanism remains unknown. The aims of this study were to identify the signaling pathways involved in IL-1β-stimulated G-CSF production and to determine whether this process was inhibited by aciculatin (8-((2R,4S,5S,6R)-tetrahydro-4,5-dihydroxy-6-methyl-2H-pyran-2-yl)-5-hydroxy-2-(4-hydroxyphenyl)-7-methoxy-4H-chromen-4-one), the major bioactive component of Chrysopogon aciculatus. IL-1β-induced cytokine expression was evaluated by measuring mRNA and protein levels by RT-PCR, ELISA, and Milliplex® assay. Whether aciculatin inhibited IL-1β-stimulated G-CSF expression, and if so, how, were evaluated using western blot assay, an electrophoretic mobility shift assay, and a reporter gene assay. Neutrophil differentiation was determined by Wright-Giemsa staining and flow cytometry. Aciculatin markedly inhibited G-CSF expression induced by IL-1β (10 ng/mL) in a concentration-dependent manner (1-10 µM). In clarifying the mechanisms involved, aciculatin was found to inhibit the IL-1β-induced activation of the IκB kinase (IKK)/IκB/nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways by suppressing the DNA binding activity of the transcription factors NF-κB and activator protein (AP)-1. Furthermore, aciculatin significantly inhibited the G-CSF-mediated phosphorylation of Janus kinase-signal transducer and activator of transcription (JAK-STAT) and Akt and neutrophil differentiation from precursor cells. Our results show that aciculatin inhibits IL-1β-stimulated G-CSF expression and the subsequent neutrophil differentiation, suggesting that it might have therapeutic potential for inflammatory arthritis.
Collapse
Affiliation(s)
- Kao-Shang Shih
- Orthopedic Department, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
- School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jyh-Horng Wang
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Wen Wu
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Che-Ming Teng
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Chih Chen
- Department of Biotechnology, Hungkuang University, Taichung, Taiwan
| | - Chia-Ron Yang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
124
|
Jackson IL, Zhang X, Hadley C, Rabbani ZN, Zhang Y, Marks S, Vujaskovic Z. Temporal expression of hypoxia-regulated genes is associated with early changes in redox status in irradiated lung. Free Radic Biol Med 2012; 53:337-46. [PMID: 22588005 PMCID: PMC3649014 DOI: 10.1016/j.freeradbiomed.2012.04.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 04/09/2012] [Accepted: 04/10/2012] [Indexed: 12/21/2022]
Abstract
The development of normal lung tissue toxicity after radiation exposure results from multiple changes in cell signaling and communication initiated at the time of the ionizing event. The onset of gross pulmonary injury is preceded by tissue hypoxia and chronic oxidative stress. We have previously shown that development of debilitating lung injury can be mitigated or prevented by administration of AEOL10150, a potent catalytic antioxidant, 24h after radiation. This suggests that hypoxia-mediated signaling pathways may play a role in late radiation injury, but the exact mechanism remains unclear. The purpose of this study was to evaluate changes in the temporal expression of hypoxia-associated genes in irradiated mouse lung and determine whether AEOL10150 alters expression of these genes. A focused oligo array was used to establish a hypoxia-associated gene expression signature for lung tissue from sham-irradiated or irradiated mice treated with or without AEOL10150. Results were further verified by RT-PCR. Forty-four genes associated with metabolism, cell growth, apoptosis, inflammation, oxidative stress, and extracellular matrix synthesis were upregulated after radiation. Elevated expression of 31 of these genes was attenuated in animals treated with AEOL10150, suggesting that expression of a number of hypoxia-associated genes is regulated by early development of oxidative stress after radiation. Genes identified herein could provide insight into the role of hypoxic signaling in radiation lung injury, suggesting novel therapeutic targets, as well as clues to the mechanism by which AEOL10150 confers pulmonary radioprotection.
Collapse
Affiliation(s)
- Isabel L. Jackson
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
| | - Xiuwu Zhang
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710 USA
| | - Caroline Hadley
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710 USA
| | - Zahid N. Rabbani
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710 USA
| | - Yu Zhang
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710 USA
| | - Sam Marks
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710 USA
| | - Zeljko Vujaskovic
- Department of Pathology, Duke University Medical Center, Durham, NC 27710 USA
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
125
|
How do cytokines trigger genomic instability? J Biomed Biotechnol 2012; 2012:536761. [PMID: 22754280 PMCID: PMC3382994 DOI: 10.1155/2012/536761] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 04/08/2012] [Indexed: 01/05/2023] Open
Abstract
Inflammation is a double-edged sword presenting a dual effect on cancer development, from one hand promoting tumor initiation and progression and from the other hand protecting against cancer through immunosurveillance mechanisms. Cytokines are crucial components of inflammation, participating in the interaction between the cells of tumor microenvironment. A comprehensive study of the role of cytokines in the context of the inflammation-tumorigenesis interplay helps us to shed light in the pathogenesis of cancer. In this paper we focus on the role of cytokines in the development of genomic instability, an evolving hallmark of cancer.
Collapse
|
126
|
Terpos E, Asli B, Christoulas D, Brouet JC, Kastritis E, Rybojad M, Bengoufa D, Dimopoulos MA, Fermand JP. Increased angiogenesis and enhanced bone formation in patients with IgM monoclonal gammopathy and urticarial skin rash: new insight into the biology of Schnitzler syndrome. Haematologica 2012; 97:1699-703. [PMID: 22689688 DOI: 10.3324/haematol.2012.067306] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Schnitzler syndrome is a rare plasma cell disorder the pathogenesis of which is still not fully understood. We evaluated the circulating levels of four major angiogenic cytokines (VEGF, angiogenin, angiopoietin-1 and angiopoietin-2) and six bone remodeling markers (sRANKL, osteoprotegerin, dickkopf-1, CTX, osteocalcin and bone-specific alkaline phosphatase-bALP) in 13 patients with Schnitzler syndrome. At diagnosis, patients had elevated angiogenic cytokines. The mean VEGF levels were almost 3.5-fold higher in Schnitzler syndrome compared to controls, while 10 of 13 patients had higher VEGF than the upper control value. Successful treatment led to a significant reduction in VEGF. Patients with Schnitzler syndrome had increased bone formation (high bALP, osteocalcin and osteoprotegerin) which was not balanced by an increase in bone resorption (normal CTX and sRANKL). These data support a role for VEGF as a new minor criterion in the diagnosis and follow up of Schnitzler syndrome, while the uncoupling of bone remodeling in favor of bone formation justifies the presence of bone densification.
Collapse
Affiliation(s)
- Evangelos Terpos
- Department of Clinical Therapeutics, University of Athens School of Medicine, Athens, Greece. /
| | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Kockar F, Yildrim H, Sagkan RI, Hagemann C, Soysal Y, Anacker J, Hamza AA, Vordermark D, Flentje M, Said HM. Hypoxia and cytokines regulate carbonic anhydrase 9 expression in hepatocellular carcinoma cells in vitro. World J Clin Oncol 2012; 3:82-91. [PMID: 22724087 PMCID: PMC3380102 DOI: 10.5306/wjco.v3.i6.82] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 05/30/2012] [Accepted: 06/05/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the expression of carbonic anhydrase (CA) 9 in human hepatocellular carcinoma (HCC) cells.
METHODS: We studied CA9 protein, CA9 mRNA and hypoxia-inducible factor-1 alpha (HIF-1α) protein levels in Hep3B cells exposed in different parallel approaches. In one of these approaches, HCC cells were exposed to extreme in vitro hypoxia (24 h 0.1% O2) without or with interleukin (IL)-1, IL-6, tumor necrosis factor-alpha (TNF-α) and transforming growth factor-beta (TGF-β) stimulation for the same hypoxic exposure time or exposed to normoxic oxygenation conditions without or with cytokine stimulation.
RESULTS: The tumour cell line analysed showed a strong hypoxic CA9 mRNA expression pattern in response to prolonged severe hypoxia with cell-line specific patterns and a marked induction of CA9 protein in response to severe hypoxia. These results were paralleled by the results for HIF-1α protein under identical oxygenation conditions with a similar expression tendency to that displayed during the CA9 protein expression experimental series. Continuous stimulation with the cytokines, IL-1, IL-6, TNF-α and TGF-β, under normoxic conditions significantly increased the carbonic anhydrase 9 expression level at both the protein and mRNA level, almost doubling the CA9 mRNA and CA9 and HIF-1α protein expression levels found under hypoxia. The findings from these experiments indicated that hypoxia is a positive regulator of CA9 expression in HCC, and the four signal transduction pathways, IL-1, IL-6, TNF-α and TGF-β, positively influence CA9 expression under both normoxic and hypoxic conditions.
CONCLUSION: These findings may potentially be considered in the design of anti- cancer therapeutic approaches involving hypoxia-induced or cytokine stimulatory effects on expression. In addition, they provide evidence of the stimulatory role of the examined cytokine families resulting in an increase in CA9 expression under different oxygenation conditions in human cancer, especially HCC, and on the role of the CA9 gene as a positive disease regulator in human cancer.
Collapse
Affiliation(s)
- Feray Kockar
- Feray Kockar, Hatice Yildrim, Department of Biology, Faculty of Art and Science, Balikesir University, 10145 Balikesir, Turkey
| | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Verma D, Bivik C, Farahani E, Synnerstad I, Fredrikson M, Enerbäck C, Rosdahl I, Söderkvist P. Inflammasome polymorphisms confer susceptibility to sporadic malignant melanoma. Pigment Cell Melanoma Res 2012; 25:506-13. [PMID: 22524199 DOI: 10.1111/j.1755-148x.2012.01008.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genetic variants of NLRP3 and NLRP1 are known to modulate levels of pro-inflammatory cytokine interleukin (IL)-1β. The purpose of this study was to investigate the association of NLRP3/NLRP1 polymorphisms with susceptibility and clinical features of malignant melanoma in a Swedish case-control study. Common variants in NLRP3/NLRP1 were investigated in sporadic malignant melanoma patients and healthy controls followed by analysis using logistic regression. NLRP3 variant (rs35829419) was significantly more common in male patients than in controls (OR, 2.22; CI, 1.27-3.86). Upon stratification, significant association with nodular melanoma was observed (OR, 2.89; CI, 1.33-6.30), which intensified in male patients (OR 4.03, CI 1.40-11.59). The NLRP1 variant (rs12150220) was significantly more common in fair-skinned female patients (OR, 1.85; CI, 1.04-3.33) and showed strong associations with nodular melanoma (OR, 6.03; CI, 1.33-25). Our data suggest that NLRP3/NLRP1 polymorphisms are associated with melanoma susceptibility; these findings warrant validation in other independent populations.
Collapse
Affiliation(s)
- Deepti Verma
- Division of Cell, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
129
|
Fitzpatrick LE, Lisovsky A, Sefton MV. The expression of sonic hedgehog in diabetic wounds following treatment with poly(methacrylic acid-co-methyl methacrylate) beads. Biomaterials 2012; 33:5297-307. [PMID: 22541537 DOI: 10.1016/j.biomaterials.2012.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 04/01/2012] [Indexed: 12/15/2022]
Abstract
The expression of native sonic hedgehog (Shh) was significantly increased in poly(methacrylic acid-co-methyl methacrylate) bead (MAA) treated wounds at day 4 compared to both poly(methyl methacrylate) bead (PMMA) treated and untreated wounds in diabetic db/db mice. MAA beads also increased the expression of the Shh transcription factor Gli3 at day 4. Previously, topical application of MAA beads (45 mol % methacrylic acid) improved wound closure and blood vessel density in excisional wounds in these mice, while PMMA beads did not. Gene expression within the granulation tissue of healing wounds was studied to provide insight into the mechanism of vessel formation and wound healing in the presence of MAA beads. In addition to the increased expression of Shh, MAA-treated wounds had increased expression of osteopontin (OPN), IL-1β and TNF-α, (at day 7) similar to the previously reported MAA response of macrophage-like and endothelial cells in vitro.
Collapse
Affiliation(s)
- Lindsay E Fitzpatrick
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada.
| | | | | |
Collapse
|
130
|
Prostanoids in tumor angiogenesis: therapeutic intervention beyond COX-2. Trends Mol Med 2012; 18:233-43. [PMID: 22425675 DOI: 10.1016/j.molmed.2012.02.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 02/10/2012] [Accepted: 02/14/2012] [Indexed: 02/06/2023]
Abstract
Prostanoids regulate angiogenesis in carcinoma and chronic inflammatory disease progression. Although prostanoid biosynthetic enzymes and signaling have been extensively analyzed in inflammation, little is known about how prostanoids mediate tumor-induced angiogenesis. Targeted cyclooxygenase (COX)-2 inhibition in tumor, stromal and endothelial cells is an attractive antiangiogenic strategy; however, the associated cardiovascular side effects have led to the development of a new generation of nonsteroidal anti-inflammatory drugs (NSAIDs) acting downstream of COX. These agents target terminal prostanoid synthases and prostanoid receptors, which may also include several peroxisome proliferator-activated receptors (PPARs). Here, we discuss the role of prostanoids as modulators of tumor angiogenesis and how prostanoid metabolism reflects complex cell-cell crosstalk that determines tumor growth. Finally, we discuss the potential of new NSAIDs for the treatment of angiogenesis-dependent tumor development.
Collapse
|
131
|
Angiogenic activity of classical hematopoietic cytokines. Leuk Res 2012; 36:537-43. [PMID: 22386730 DOI: 10.1016/j.leukres.2012.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 01/24/2012] [Accepted: 02/07/2012] [Indexed: 12/24/2022]
Abstract
Hematopoiesis is regulated by several cytokines with pleiotropic activity. Several evidences have clearly demonstrated that these molecules, formerly regarded as specific for the hematopoietic system, also affect certain endothelial cell functions and that hematopoietic factors clearly influence angiogenesis. This review article summarizes the most important literature data concerning this inconvertible relationship.
Collapse
|
132
|
Jagielska J, Kapopara PR, Salguero G, Scherr M, Schütt H, Grote K, Schieffer B, Bavendiek U. Interleukin-1 assembles a proangiogenic signaling module consisting of caveolin-1, tumor necrosis factor receptor-associated factor 6, p38-mitogen-activated protein kinase (MAPK), and MAPK-activated protein kinase 2 in endothelial cells. Arterioscler Thromb Vasc Biol 2012; 32:1280-8. [PMID: 22345171 DOI: 10.1161/atvbaha.111.243477] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Interleukin-1β (IL-1β) is a major cytokine linking inflammation and angiogenesis in pathological vascular processes, such as atherosclerosis and tumor neoangiogenesis. However, signaling pathways mediating IL-1β-induced proangiogenic processes in endothelial cells (ECs) have barely been elucidated yet. Therefore, the present study investigated IL-1β-induced proangiogenic signaling in ECs. METHODS AND RESULTS IL-1β potently induced tube formation and migration of ECs. This was associated with and dependent on activation of p38-mitogen-activated protein kinase (MAPK) and MAPK-activated protein kinase 2 (MK2) as determined by pharmacological inhibition and gene silencing. Furthermore, silencing of the adaptor protein tumor necrosis factor receptor-associated factor 6 (TRAF6) (lentiviral short hairpin RNA) inhibited these IL-1β-induced processes. Moreover, IL-1β promoted translocation of TRAF6 to insoluble cellular fractions (containing membrane rafts/caveolae) and interaction of TRAF6 with caveolin-1. Accordingly, cellular cholesterol depletion (cyclodextrin) and silencing of caveolin-1 (small interfering RNA) inhibited IL-1β-induced activation of p38-MAPK and MK2, as well as IL-1β-induced tube formation and migration. Finally, silencing of TRAF6 and MK2 deficiency inhibited IL-1β-induced microvessel outgrowth in murine aortic rings ex vivo, and deficiency of MK2 or caveolin-1 significantly reduced IL-1β-induced angiogenesis in mice in vivo (Matrigel plug assay). CONCLUSIONS IL-1β assembles a proangiogenic signaling module consisting of caveolin-1, TRAF6, p38-MAPK, and MK2 in ECs, representing a potential target to intervene into angiogenesis-dependent processes and diseases.
Collapse
Affiliation(s)
- Joanna Jagielska
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | |
Collapse
|
133
|
Kvietys PR, Granger DN. Role of reactive oxygen and nitrogen species in the vascular responses to inflammation. Free Radic Biol Med 2012; 52:556-592. [PMID: 22154653 PMCID: PMC3348846 DOI: 10.1016/j.freeradbiomed.2011.11.002] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 11/04/2011] [Accepted: 11/04/2011] [Indexed: 12/23/2022]
Abstract
Inflammation is a complex and potentially life-threatening condition that involves the participation of a variety of chemical mediators, signaling pathways, and cell types. The microcirculation, which is critical for the initiation and perpetuation of an inflammatory response, exhibits several characteristic functional and structural changes in response to inflammation. These include vasomotor dysfunction (impaired vessel dilation and constriction), the adhesion and transendothelial migration of leukocytes, endothelial barrier dysfunction (increased vascular permeability), blood vessel proliferation (angiogenesis), and enhanced thrombus formation. These diverse responses of the microvasculature largely reflect the endothelial cell dysfunction that accompanies inflammation and the central role of these cells in modulating processes as varied as blood flow regulation, angiogenesis, and thrombogenesis. The importance of endothelial cells in inflammation-induced vascular dysfunction is also predicated on the ability of these cells to produce and respond to reactive oxygen and nitrogen species. Inflammation seems to upset the balance between nitric oxide and superoxide within (and surrounding) endothelial cells, which is necessary for normal vessel function. This review is focused on defining the molecular targets in the vessel wall that interact with reactive oxygen species and nitric oxide to produce the characteristic functional and structural changes that occur in response to inflammation. This analysis of the literature is consistent with the view that reactive oxygen and nitrogen species contribute significantly to the diverse vascular responses in inflammation and supports efforts that are directed at targeting these highly reactive species to maintain normal vascular health in pathological conditions that are associated with acute or chronic inflammation.
Collapse
Affiliation(s)
- Peter R Kvietys
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA.
| |
Collapse
|
134
|
Kasimanickam RK, Kasimanickam VR, Haldorson GJ, Tibary A. Effect of tocopherol supplementation during last trimester of pregnancy on mRNA abundances of interleukins and angiogenesis in ovine placenta and uterus. Reprod Biol Endocrinol 2012; 10:4. [PMID: 22269218 PMCID: PMC3398327 DOI: 10.1186/1477-7827-10-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 01/23/2012] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Interleukins (IL) play an important role in angiogenesis. Tocopherol possesses immunomodulating effect in addition to antioxidant property. The objective of this study was to determine whether gamma tocopherol's (gT) angiogenic activity in placental network is enhanced via promoting interleukins. METHODS Pregnant ewes (N=18) were supplemented, orally, with 500 mg of alpha tocopherol (aT; N=6) or 1,000 mg of gT (N=7) or placebo (CON; N=5) once daily from 107 to 137 days post breeding. Uterine and placental tissue samples were obtained at the end of supplementation to evaluate relative mRNA expressions of IL-1b, IL-6, IL-8, Tumor Necrosis Factor (TNF) alpha, Vascular Endothelial Growth Factor (VEGF), kinase insert domain receptor (KDR; VGFR2; a type III receptor tyrosine kinase), and soluble fms-like tyrosine kniase-1 (sFlt1 or sVEGFR1) in uterus, caruncle and cotyledon. RESULTS Oral supplementation of gT increased IL-6, IL-8, KDR and VEGF mRNA abundances whereas sFlt1 mRNA abundance was suppressed in uterus, caruncle and cotyledon, compared to aT and placebo treated ewes (P<0.05). The TNF alpha and IL-1b mRNA abundances were suppressed in uterus, caruncle and cotyledon but TNF alpha is higher in gT group compared to aT group (P<0.05), whereas IL-1b was similar between treatment groups (P>0.1). CONCLUSIONS Gamma tocopherol supplementation increased IL-6, IL-8, and KDR mRNA abundances and suppressed sFlt1 and TNFalpha mRNA abundances thereby increased VEGF mRNA expression and angiogenesis in placental vascular network during late gestation. It is plausible that the angiogenic effect of gamma tocopherol in placental vascular network is exerted via an alternate path by enhancing IL-6 and IL-8.
Collapse
Affiliation(s)
| | | | - Gary J Haldorson
- College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Ahmed Tibary
- College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
135
|
Fontanella M, Rubino E, Crobeddu E, Gallone S, Gentile S, Garbossa D, Ducati A, Pinessi L, Rainero I. Brain Arteriovenous Malformations Are Associated With Interleukin-1 Cluster Gene Polymorphisms. Neurosurgery 2012; 70:12-7. [DOI: 10.1227/neu.0b013e31822d9881] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
136
|
The p38 mitogen-activated protein kinases modulate endothelial cell survival and tissue repair. Inflamm Res 2011; 61:233-44. [PMID: 22138711 DOI: 10.1007/s00011-011-0405-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 10/17/2011] [Accepted: 11/11/2011] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE AND DESIGN This study is designed to investigate the role of p38 MAPK in modulating human pulmonary artery endothelial cells (HPAECs) survival and tissue repair functions. METHODS HPAECs (passage 8-12) were used for all experiments. Cells were treated with IL-1β (0.5 or 2 ng/ml) or p38 inhibitor (SB203580 or SB220025, 5 μM each). Cells were also transfected with 50 nM siRNAs. Cell length was measured using ImageJ software. Collagen gel contraction and wound close assay were performed to evaluate tissue repair functions. RESULTS IL-1β activated p38 MAPK and induced morphologic change of HPAECs. The p38 inhibitors further augmented IL-1β-induced cell morphologic change, prevented cell death, and augmented collagen gel contraction. Suppression of p38α, γ, or δ, but not p38β resulted in cell morphologic alteration, and suppressing any one of p38 isoforms by siRNAs increased cell survival. Suppression of p38α or δ augmented gel contraction. While p38α suppression stimulated cell migration, suppressing the rest of three isoforms inhibit cell migration. Nuclear factor p65-siRNA blocked IL-1β-induced cell morphologic change, but did not affect p38 inhibitor-induced change. CONCLUSION These findings suggest that p38 MAPK may negatively modulate tissue repair functions of endothelial cells via p65 independent pathway.
Collapse
|
137
|
Germano G, Mantovani A, Allavena P. Targeting of the innate immunity/inflammation as complementary anti-tumor therapies. Ann Med 2011; 43:581-93. [PMID: 21756064 DOI: 10.3109/07853890.2011.595732] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Different types of cancer take advantage of inflammatory components to improve their life-span in the organs. A sustenance of growth factors and cytokines (e.g. interleukin (IL)-1, tumor necrosis factor, IL-6, vascular endothelial growth factor) supports malignant cell progression and contributes to suppress the body immune defense. Strategies to modulate the host micro-environment offer new approaches for anti-cancer therapies. For these reasons new molecules with anti-tumor and anti-inflammatory features (e.g. trabectedin) are looked at with new eyes in the light of the crucial link between inflammation and cancer.
Collapse
Affiliation(s)
- Giovanni Germano
- Department of Immunology and Inflammation, IRCCS Humanitas Clinical Institute, Rozzano, Milan, Italy
| | | | | |
Collapse
|
138
|
Fitzpatrick LE, Chan JW, Sefton MV. On the mechanism of poly(methacrylic acid –co– methyl methacrylate)-induced angiogenesis: Gene expression analysis of dTHP-1 cells. Biomaterials 2011; 32:8957-67. [DOI: 10.1016/j.biomaterials.2011.08.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 08/09/2011] [Indexed: 01/13/2023]
|
139
|
Rider P, Carmi Y, Guttman O, Braiman A, Cohen I, Voronov E, White MR, Dinarello CA, Apte RN. IL-1α and IL-1β recruit different myeloid cells and promote different stages of sterile inflammation. THE JOURNAL OF IMMUNOLOGY 2011; 187:4835-43. [PMID: 21930960 DOI: 10.4049/jimmunol.1102048] [Citation(s) in RCA: 396] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The immune system has evolved to protect the host from invading pathogens and to maintain tissue homeostasis. Although the inflammatory process involving pathogens is well documented, the intrinsic compounds that initiate sterile inflammation and how its progression is mediated are still not clear. Because tissue injury is usually associated with ischemia and the accompanied hypoxia, the microenvironment of various pathologies involves anaerobic metabolites and products of necrotic cells. In the current study, we assessed in a comparative manner the role of IL-1α and IL-1β in the initiation and propagation of sterile inflammation induced by products of hypoxic cells. We found that following hypoxia, the precursor form of IL-1α, and not IL-1β, is upregulated and subsequently released from dying cells. Using an inflammation-monitoring system consisting of Matrigel mixed with supernatants of hypoxic cells, we noted accumulation of IL-1α in the initial phase, which correlated with the infiltration of neutrophils, and the expression of IL-1β correlated with later migration of macrophages. In addition, we were able to show that IL-1 molecules from cells transfected with either precursor IL-1α or mature IL-1β can recruit neutrophils or macrophages, respectively. Taken together, these data suggest that IL-1α, released from dying cells, initiates sterile inflammation by inducing recruitment of neutrophils, whereas IL-1β promotes the recruitment and retention of macrophages. Overall, our data provide new insight into the biology of IL-1 molecules as well as on the regulation of sterile inflammation.
Collapse
Affiliation(s)
- Peleg Rider
- The Shraga Segal Department of Microbiology and Immunology and The Cancer Research Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Goyal S, Chauhan SK, Dana R. Blockade of prolymphangiogenic vascular endothelial growth factor C in dry eye disease. ACTA ACUST UNITED AC 2011; 130:84-9. [PMID: 21911653 DOI: 10.1001/archophthalmol.2011.266] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To determine whether blocking prolymphangiogenic factors such as vascular endothelial growth factor C (VEGF-C) would suppress alloimmunity in dry eye disease using a murine model. METHODS The effects of intraperitoneal injections of 400 μg of anti-VEGF-C antibody (treated group) and intraperitoneal normal saline (untreated group) were studied in murine dry eyes induced by exposing mice to high-flow desiccated air in a controlled-environment chamber. Growth of lymphatic vessels and infiltration of macrophages were evaluated by immunohistochemistry using CD31 (panendothelial marker), lymphatic vessel endothelial receptor 1 (lymphatic endothelial marker), and CD11b (monocyte and macrophage marker). Real-time polymerase chain reaction was performed to quantify expression of different inflammatory cytokine transcripts in the conjunctiva and lymph nodes as well as vascular endothelial growth factors and their receptors (VEGF-A, VEGF-C, VEGF-D, VEGFR-2, and VEGFR-3) in the cornea. RESULTS Blocking VEGF-C led to significant reductions in lymphatic caliber (P = .02) and lymphatic area (P = .006) in the corneas of mice with dry eye disease. In addition to significantly decreasing CD11b(+) cells (P = .005), anti-VEGF-C treatment significantly decreased transcript levels of VEGF-C (P = .002), VEGF-D (P = .01), and VEGFR-3 (P = .02) in the corneas of the treated group. A significant decrease in expression of inflammatory cytokines in the conjunctiva (interleukin 1α, P = .003; interleukin 1β, P = .02; interleukin 6, P = .005) and lymph nodes (interferon γ, P = .008; interleukin 17, P = .003) was also seen with anti-VEGF-C treatment. CONCLUSION Treatment with anti-VEGF-C led to significant improvement in dry eye disease, reflected by a decrease in inflammation at the clinical, molecular, and cellular levels. CLINICAL RELEVANCE Targeting prolymphangiogenic growth factors or their receptors could inhibit the trafficking of antigen-presenting cells to the draining lymph nodes and hence prove to be a potential therapeutic target for dry eye disease.
Collapse
Affiliation(s)
- Sunali Goyal
- Schepens Eye Research Institute, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
141
|
Abstract
Th17 cells, in addition to their proinflammatory functions, have been recognized as potent inducers of angiogenesis in autoimmune diseases and malignancies. In the present study, we demonstrate distinct mechanisms by which IL-17 induces lymphangiogenesis. Using the mouse cornea micropocket and cell culture assays, our data demonstrate that IL-17 directly promotes growth of lymphatic vessels by inducing increased expression of prolymphangiogenic VEGF-D and proliferation of lymphatic endothelial cells. However, IL-17-induced growth of blood vessels is primarily mediated through IL-1β secretion by IL-17-responsive cells. Furthermore, in vivo blockade of IL-17 in a preclinical model of Th17-dominant autoimmune ocular disease demonstrates a significant reduction in the corneal lymphangiogenesis and in the progression of clinical disease. Taken together, our findings demonstrate a novel prolymphangiogenic function for Th17/IL-17, indicating that IL-17 can promote the progression and amplification of immunity in part through its induction of lymphangiogenesis.
Collapse
|
142
|
Georgi MK, Vigilance J, Dewar AM, Frame MD. Terminal arteriolar network structure/function and plasma cytokine levels in db/db and ob/ob mouse skeletal muscle. Microcirculation 2011; 18:238-51. [PMID: 21418374 DOI: 10.1111/j.1549-8719.2011.00084.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To investigate the terminal arteriolar network structure and function in relation to circulating plasma cytokine levels in db/db, ob/ob, and their genetic background control, C57/bl6, mice. METHODS Arteriolar network size and erythrocyte distribution were observed in the resting cremaster muscle (n = 45, pentobarbital 50 mg/kg i.p.). Structural remodeling and inflammatory state were related to 21 plasma cytokine levels. RESULTS db/db networks were shorter, had fewer branches, and smaller diameters than C57/bl6 controls. ob/ob networks were longer, with similar branch numbers, however with non-uniform diameters. Shunting of erythrocytes to the specific terminal arteriolar branches of the network (functional rarefaction) was prominent in db/db and ob/ob, with further evidence of shunting between networks seen as no flow to 50% of ob/ob arteriolar networks. CONCLUSIONS Altered levels of plasma cytokines are consistent with structural remodeling seen in db/db, and a pro-inflammatory state for both db/db and ob/ob. Differences in network structure alone predict overall reduced uniform oxygen delivery in db/db or ob/ob. Shunting probably increases heterogeneous oxygen delivery and is strain-dependent.
Collapse
Affiliation(s)
- Melissa K Georgi
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794-5281, USA
| | | | | | | |
Collapse
|
143
|
Lavalette S, Raoul W, Houssier M, Camelo S, Levy O, Calippe B, Jonet L, Behar-Cohen F, Chemtob S, Guillonneau X, Combadière C, Sennlaub F. Interleukin-1β inhibition prevents choroidal neovascularization and does not exacerbate photoreceptor degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2416-23. [PMID: 21514452 DOI: 10.1016/j.ajpath.2011.01.013] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 12/20/2010] [Accepted: 01/13/2011] [Indexed: 01/30/2023]
Abstract
The pro-inflammatory cytokine IL-1β has been shown to promote angiogenesis. It can have a neurotoxic or neuroprotective effect. Here, we have studied the expression of IL-1β in vivo and the effect of the IL-1 receptor antagonist on choroidal neovascularization (CNV) and retinal degeneration (RD). IL-1β expression significantly increased after laser injury (real time PCR) in C57BL/6 mice, in the C57BL/6 Cx3cr1(-/-) model of age-related macular degeneration (enzyme-linked immunoabsorbent assay), and in albino Wistar rats and albino BALB Cx3cr1(+/+) and Cx3cr1(-/-) mice (enzyme-linked immunoabsorbent assay) after light injury. IL-1β was localized to Ly6G-positive, Iba1-negative infiltrating neutrophils in laser-induced CNV as determined by IHC. IL-1 receptor antagonist treatment significantly inhibited CNV but did not affect Iba1-positive macrophage recruitment to the injury site. IL-1β significantly increased endothelial cell outgrowth in aortic ring assay independently of vascular endothelial growth factor, suggesting a direct effect of IL-1β on choroidal endothelial cell proliferation. Inhibition of IL-1β in light- and laser-induced RD models did not alter photoreceptor degeneration in Wistar rats, C57BL/6 mice, or RD-prone Cx3cr1(-/-) mice. Our results suggest that IL-1β inhibition might represent a valuable and safe alternative to inhibition of vascular endothelial growth factor in the control of CNV in the context of concomitant photoreceptor degeneration as observed in age-related macular degeneration.
Collapse
Affiliation(s)
- Sophie Lavalette
- INSERM, UMR S 872, Centre de Recherche des Cordeliers, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Engineering of vascularized adipose constructs. Cell Tissue Res 2011; 347:747-57. [DOI: 10.1007/s00441-011-1226-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 07/22/2011] [Indexed: 12/19/2022]
|
145
|
Zhou TB, Yang GS. Roles of vascular endothelial growth factor in acute rejection reaction following liver transplantation. Transpl Immunol 2011; 25:207-9. [PMID: 21856421 DOI: 10.1016/j.trim.2011.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Accepted: 08/05/2011] [Indexed: 12/20/2022]
Abstract
The presently known cytokines that participate in acute rejection of organ transplantation include four categories by order of function: inflammatory cytokines, immunospecific cytokines, inflammatory cell activating cytokines and growth cytokines. Of them, growth cytokines that directly induce division, proliferation and migration of endothelial cells mainly include the vascular endothelial growth factor (VEGF) family and the fibroblast growth factor (FGF) family [1]. Recent studies [2] showed that interactions and time overlap of inflammatory cell infiltration and angiogenesis are the main mechanisms that induce acute rejection (AR) following organ transplantation, which has been demonstrated by the clinical fact that AR symptoms after liver transplantation could only be relieved by combination use of drugs for improving micro vessels and those for improving micro bile ducts. This article is a review of VEGF that mediates inflammatory cell infiltration and angiogenesis in the portal area [3].
Collapse
Affiliation(s)
- Tian-Bao Zhou
- The Hepatic Surgery Center, The Affiliated Ningbo No. 2 Hospital, Ningbo University School of Medicine, Ningbo, China.
| | | |
Collapse
|
146
|
Carbone C, Moccia T, Zhu C, Paradiso G, Budillon A, Chiao PJ, Abbruzzese JL, Melisi D. Anti-VEGF treatment-resistant pancreatic cancers secrete proinflammatory factors that contribute to malignant progression by inducing an EMT cell phenotype. Clin Cancer Res 2011; 17:5822-32. [PMID: 21737511 DOI: 10.1158/1078-0432.ccr-11-1185] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE The resistance of tumors to antiangiogenic therapies is becoming increasingly relevant. There are currently no validated predictive biomarkers for selecting which cancer patients will benefit from antiangiogenic therapy. Also lacking are resistance biomarkers that can identify which escape pathways should be targeted after tumors develop resistance to VEGF treatment. Recent studies showed that anti-VEGF treatment can make tumor cells more aggressive and metastatic. However, the mechanisms and mediators of this are unidentified. Therefore, we aimed this study at directly identifying the tumor cell-initiated mechanisms responsible for the resistance of pancreatic cancer to anti-VEGF treatment. EXPERIMENTAL DESIGN We established and validated two murine models of human pancreatic cancer resistant to the VEGF-specific antibody bevacizumab in vivo. We used a genome-wide analysis to directly identify which tumor-secreted factors were overexpressed by pancreatic cancer cells that were resistant to anti-VEGF treatment. RESULTS Rather than direct proangiogenic factors, we identified several proinflammatory factors that were expressed at higher levels in cells resistant to anti-VEGF treatment than in treatment-sensitive control cells. These proinflammatory factors acted in a paracrine manner to stimulate the recruitment of CD11b(+) proangiogenic myeloid cells. Also, we found that secreted factors overexpressed by anti-VEGF treatment-resistant pancreatic cancer cells acted in an autocrine manner to induce epithelial-to-mesenchymal transition (EMT) and were thus responsible for increased aggressiveness of bevacizumab-resistant pancreatic tumors. CONCLUSIONS Our results identified proinflammatory factors and EMT markers as potential biomarkers for selecting patients with pancreatic cancer for antiangiogenic therapy.
Collapse
Affiliation(s)
- Carmine Carbone
- Experimental Pharmacology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori-Fondazione G. Pascale, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Ohno-Matsui K. Parallel findings in age-related macular degeneration and Alzheimer’s disease. Prog Retin Eye Res 2011; 30:217-38. [DOI: 10.1016/j.preteyeres.2011.02.004] [Citation(s) in RCA: 186] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Revised: 02/24/2011] [Accepted: 02/24/2011] [Indexed: 12/16/2022]
|
148
|
Ovarian epithelial-stromal interactions: role of interleukins 1 and 6. Obstet Gynecol Int 2011; 2011:358493. [PMID: 21765834 PMCID: PMC3135012 DOI: 10.1155/2011/358493] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 04/12/2011] [Accepted: 04/27/2011] [Indexed: 01/03/2023] Open
Abstract
Ovarian epithelial cancer is the most lethal gynecologic malignancy. The high mortality is attributed to the fact that most cases typically present in late stage when ovarian cancer (OC) has already spread beyond the ovary. Ovarian epithelial cancer cells are shed into intraperitoneal ascites and easily disseminate throughout the peritoneal cavity with preferential metastasis to the omentum, peritoneum, and local organs. Understanding how ovarian epithelial cells interact with and modulate their microenvironment can provide insight into the molecular mechanism(s) involved with malignant transformation and progression which may eventually identify novel diagnostic, prognostic, and therapeutic targets. The objective of this paper is to provide a brief consideration of ovarian surface epithelial-stromal interactions in regard to normal physiological function and tumor progression as influenced by two potentially key interleukins, interleukins-1 (IL-1) and -6 (IL-6), present in the microenvironment. Lastly, we will consider the clinical implications of IL-1 and IL-6 for OC patients.
Collapse
|
149
|
Interleukin-33: a novel player in osteonecrosis of the femoral head? Joint Bone Spine 2011; 78:550-4. [PMID: 21703905 DOI: 10.1016/j.jbspin.2011.04.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 04/20/2011] [Indexed: 12/21/2022]
Abstract
Osteonecrosis of the femoral head (ONFH) is a disabling disease affecting young adults, which usually leads to the destruction of the hip joint. It is mainly due to an inadequate blood supply that causes the death of osteocytes and bone marrow cells. Joint salvaging procedures are numerous but relatively inefficient, justifying the need for new therapeutic strategies. In this regard, the recently discovered interleukin (IL)-33 alarmin appears as a possible target. Indeed, IL-33 seems to be specifically released by necrotic cells, and interestingly, is constitutively expressed in human bone, in particular by osteocytes, osteoblasts and marrow adipocytes. Moreover, recent reports suggesting that IL-33 modulates angiogenesis, vascular permeability, osteoclastogenesis and bone resorption, indicate that IL-33 may play a role in ONFH.
Collapse
|
150
|
Immature myeloid cells accumulate in mouse placenta and promote angiogenesis. Am J Obstet Gynecol 2011; 204:544.e18-23. [PMID: 21420066 DOI: 10.1016/j.ajog.2011.01.060] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Revised: 01/25/2011] [Accepted: 01/31/2011] [Indexed: 12/17/2022]
Abstract
OBJECTIVE We sought to determine whether CD11b(+)Gr1(+) immature myeloid cells (IMCs), which have been shown to promote tumor angiogenesis, accumulate in the placenta and similarly contribute to blood vessel formation. STUDY DESIGN Experiments were performed on 6- to 8-week-old C57Bl/6J female mice. Placentas from pregnant mice or B16F10 tumors that were subcutaneously implanted were analyzed by flow cytometry and confocal microscopy. To determine the proangiogenic potential of IMCs, Matrigel plug assays were performed. RESULTS IMCs infiltrate the placenta in the proximity of blood vessels, reaching peak concentration at midpregnancy. When isolated from either placentas or B16F10 melanoma tumors, IMCs actively promoted endothelial cell migration into Matrigel plugs in vivo. Furthermore, placental IMCs, similar to tumor-derived IMCs, expressed matrix metalloproteinase-9 and Bv8, 2 pivotal proangiogenic proteins. CONCLUSION IMCs that express matrix metalloproteinase-9 and Bv8 infiltrate placentas of pregnant mice and actively promote angiogenesis. These cells show striking similarity to IMCs that populate malignant tumors.
Collapse
|